Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Neurol ; 338: 113607, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33460644

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) is a common dose-limiting side effect of cancer treatment, often associated with degeneration of sensory axons or their terminal regions. Presence of the slow Wallerian degeneration protein (WLDS), or genetic deletion of sterile alpha and TIR motif containing protein 1 (SARM1), which strongly protect axons from degeneration after injury or axonal transport block, alleviate pain in several CIPN models. However, oxaliplatin can cause an acute pain response, suggesting a different mechanism of pain generation. Here, we tested whether the presence of WLDS or absence of SARM1 protects against acute oxaliplatin-induced pain in mice after a single oxaliplatin injection. In BL/6 and WldS mice, oxaliplatin induced significant mechanical and cold hypersensitivities which were absent in Sarm1-/- mice. Despite the presence of hypersensitivity there was no significant loss of intraepidermal nerve fibers (IENFs) in the footpads of any mice after oxaliplatin treatment, suggesting that early stages of pain hypersensitivity could be independent of axon degeneration. To identify other changes that could underlie the pain response, RNA sequencing was carried out in DRGs from treated and control mice of each genotype. Sarm1-/- mice had fewer gene expression changes than either BL/6 or WldS mice. This is consistent with the pain measurements in demonstrating that Sarm1-/- DRGs remain relatively unchanged after oxaliplatin treatment, unlike those in BL/6 and WldS mice. Changes in levels of four transcripts - Alas2, Hba-a1, Hba-a2, and Tfrc - correlated with oxaliplatin-induced pain, or absence thereof, across the three genotypes. Our findings suggest that targeting SARM1 could be a viable therapeutic approach to prevent oxaliplatin-induced acute neuropathic pain.


Asunto(s)
Antineoplásicos/toxicidad , Proteínas del Dominio Armadillo/metabolismo , Proteínas del Citoesqueleto/metabolismo , Neuralgia/inducido químicamente , Neuralgia/metabolismo , Oxaliplatino/toxicidad , Animales , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
3.
Gut ; 68(4): 633-644, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29483303

RESUMEN

OBJECTIVE: Integration of nutritional, microbial and inflammatory events along the gut-brain axis can alter bowel physiology and organism behaviour. Colonic sensory neurons activate reflex pathways and give rise to conscious sensation, but the diversity and division of function within these neurons is poorly understood. The identification of signalling pathways contributing to visceral sensation is constrained by a paucity of molecular markers. Here we address this by comprehensive transcriptomic profiling and unsupervised clustering of individual mouse colonic sensory neurons. DESIGN: Unbiased single-cell RNA-sequencing was performed on retrogradely traced mouse colonic sensory neurons isolated from both thoracolumbar (TL) and lumbosacral (LS) dorsal root ganglia associated with lumbar splanchnic and pelvic spinal pathways, respectively. Identified neuronal subtypes were validated by single-cell qRT-PCR, immunohistochemistry (IHC) and Ca2+-imaging. RESULTS: Transcriptomic profiling and unsupervised clustering of 314 colonic sensory neurons revealed seven neuronal subtypes. Of these, five neuronal subtypes accounted for 99% of TL neurons, with LS neurons almost exclusively populating the remaining two subtypes. We identify and classify neurons based on novel subtype-specific marker genes using single-cell qRT-PCR and IHC to validate subtypes derived from RNA-sequencing. Lastly, functional Ca2+-imaging was conducted on colonic sensory neurons to demonstrate subtype-selective differential agonist activation. CONCLUSIONS: We identify seven subtypes of colonic sensory neurons using unbiased single-cell RNA-sequencing and confirm translation of patterning to protein expression, describing sensory diversity encompassing all modalities of colonic neuronal sensitivity. These results provide a pathway to molecular interrogation of colonic sensory innervation in health and disease, together with identifying novel targets for drug development.


Asunto(s)
Colon/inervación , Células Receptoras Sensoriales/clasificación , Análisis de Secuencia de ARN , Transcriptoma , Animales , Inmunohistoquímica , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Aging Cell ; 17(4): e12795, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29943484

RESUMEN

Despite pain prevalence altering with age, the effects of aging on the properties of nociceptors are not well understood. Nociceptors, whose somas are located in dorsal root ganglia, are frequently divided into two groups based on their ability to bind isolectin B4 (IB4). Here, using cultured neurons from 1-, 3-, 5-, 8-, 12-, and 18-month-old mice, we investigate age-dependent changes in IB4-positive and IB4-negative neurons. Current-clamp experiments at physiological temperature revealed nonlinear changes in firing frequency of IB4-positive, but not IB4-negative neurons, with a peak at 8 months. This was likely due to the presence of proexcitatory conductances activated at depolarized membrane potentials and significantly higher input resistances found in IB4-positive neurons from 8-month-old mice. Repetitive firing in nociceptors is driven primarily by the TTX-resistant sodium current, and indeed, IB4-positive neurons from 8-month-old mice were found to receive larger contributions from the TTX-resistant window current around the resting membrane potential. To further address the mechanisms behind these differences, we performed RNA-seq experiments on IB4-positive and IB4-negative neurons from 1-, 8-, and 18-month-old mice. We found a larger number of genes significantly affected by age within the IB4-positive than IB4-negative neurons from 8-month-old mice, including known determinants of nociceptor excitability. The above pronounced age-dependent changes at the cellular and molecular levels in IB4-positive neurons point to potential mechanisms behind the reported increase in pain sensitivity in middle-aged rodents and humans, and highlight the possibility of targeting a particular group of neurons in the development of age-tailored pain treatments.


Asunto(s)
Senescencia Celular/genética , Glicoproteínas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Nociceptores/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica/genética , Glicoproteínas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/citología , Debilidad Muscular/genética , Nociceptores/citología
5.
Nat Biotechnol ; 36(4): 328-337, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29553577

RESUMEN

Age-related macular degeneration (AMD) remains a major cause of blindness, with dysfunction and loss of retinal pigment epithelium (RPE) central to disease progression. We engineered an RPE patch comprising a fully differentiated, human embryonic stem cell (hESC)-derived RPE monolayer on a coated, synthetic basement membrane. We delivered the patch, using a purpose-designed microsurgical tool, into the subretinal space of one eye in each of two patients with severe exudative AMD. Primary endpoints were incidence and severity of adverse events and proportion of subjects with improved best-corrected visual acuity of 15 letters or more. We report successful delivery and survival of the RPE patch by biomicroscopy and optical coherence tomography, and a visual acuity gain of 29 and 21 letters in the two patients, respectively, over 12 months. Only local immunosuppression was used long-term. We also present the preclinical surgical, cell safety and tumorigenicity studies leading to trial approval. This work supports the feasibility and safety of hESC-RPE patch transplantation as a regenerative strategy for AMD.


Asunto(s)
Células Madre Embrionarias Humanas/trasplante , Degeneración Macular/terapia , Epitelio Pigmentado de la Retina/trasplante , Agudeza Visual/fisiología , Anciano , Animales , Membrana Basal/diagnóstico por imagen , Membrana Basal/crecimiento & desarrollo , Diferenciación Celular/genética , Femenino , Humanos , Degeneración Macular/diagnóstico por imagen , Degeneración Macular/patología , Masculino , Ratones , Persona de Mediana Edad , Epitelio Pigmentado de la Retina/diagnóstico por imagen , Epitelio Pigmentado de la Retina/crecimiento & desarrollo , Trasplante de Células Madre/efectos adversos , Porcinos , Tomografía de Coherencia Óptica
6.
SLAS Discov ; 23(6): 585-596, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29547351

RESUMEN

Screening against a disease-relevant phenotype to identify compounds that change the outcome of biological pathways, rather than just the activity of specific targets, offers an alternative approach to find modulators of disease characteristics. However, in pain research, use of in vitro phenotypic screens has been impeded by the challenge of sourcing relevant neuronal cell types in sufficient quantity and developing functional end-point measurements with a direct disease link. To overcome these hurdles, we have generated human induced pluripotent stem cell (hiPSC)-derived sensory neurons at a robust production scale using the concept of cryopreserved "near-assay-ready" cells to decouple complex cell production from assay development and screening. hiPSC sensory neurons have then been used for development of a 384-well veratridine-evoked calcium flux assay. This functional assay of neuronal excitability was validated for phenotypic relevance to pain and other hyperexcitability disorders through screening a small targeted validation compound subset. A 2700-compound chemogenomics screen was then conducted to profile the range of target-based mechanisms able to inhibit veratridine-evoked excitability. This report presents the assay development, validation, and screening data. We conclude that high-throughput-compatible pain-relevant phenotypic screening with hiPSC sensory neurons is feasible and ready for application for the identification of new targets, pathways, mechanisms of action, and compounds for modulating neuronal excitability.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Dolor/patología , Células Receptoras Sensoriales/citología , Células Cultivadas , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Fenotipo
7.
Br J Pharmacol ; 175(12): 2272-2283, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29150838

RESUMEN

BACKGROUND AND PURPOSE: TREK two-pore-domain potassium (K2P ) channels play a critical role in regulating the excitability of somatosensory nociceptive neurons and are important mediators of pain perception. An understanding of the roles of TREK channels in pain perception and, indeed, in other pathophysiological conditions, has been severely hampered by the lack of potent and/or selective activators and inhibitors. In this study, we describe a new, selective opener of TREK channels, GI-530159. EXPERIMENTAL APPROACH: The effect of GI-530159 on TREK channels was demonstrated using 86 Rb efflux assays, whole-cell and single-channel patch-clamp recordings from recombinant TREK channels. The expression of K2P 2.1 (TREK1), K2P 10.1 (TREK2) and K2P 4.1 (TRAAK) channels was determined using transcriptome analysis from single dorsal root ganglion (DRG) cells. Current-clamp recordings from cultured rat DRG neurons were used to measure the effect of GI-530159 on neuronal excitability. KEY RESULTS: For recombinant human TREK1 channels, GI-530159 had similar low EC50 values in Rb efflux experiments and electrophysiological recordings. It activated TREK2 channels, but it had no detectable action on TRAAK channels nor any significant effect on other K channels tested. Current-clamp recordings from cultured rat DRG neurones showed that application of GI-530159 at 1 µM resulted in a significant reduction in firing frequency and a small hyperpolarization of resting membrane potential. CONCLUSIONS AND IMPLICATIONS: This study provides pharmacological evidence for the presence of mechanosensitive TREK K2P channels in sensory neurones and suggests that development of selective K2P channel openers like GI-530159 could aid in the development of novel analgesic agents. LINKED ARTICLES: This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.


Asunto(s)
Ganglios Espinales/efectos de los fármacos , Neuronas/efectos de los fármacos , Canales de Potasio de Dominio Poro en Tándem/agonistas , Animales , Células CHO , Línea Celular , Cricetulus , Relación Dosis-Respuesta a Droga , Ganglios Espinales/metabolismo , Células HEK293 , Humanos , Estructura Molecular , Neuronas/metabolismo , Ratas , Relación Estructura-Actividad
8.
Nat Genet ; 50(1): 54-61, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29229984

RESUMEN

Induced pluripotent stem cells (iPSCs), and cells derived from them, have become key tools for modeling biological processes, particularly in cell types that are difficult to obtain from living donors. Here we present a map of regulatory variants in iPSC-derived neurons, based on 123 differentiations of iPSCs to a sensory neuronal fate. Gene expression was more variable across cultures than in primary dorsal root ganglion, particularly for genes related to nervous system development. Using single-cell RNA-sequencing, we found that the number of neuronal versus contaminating cells was influenced by iPSC culture conditions before differentiation. Despite high differentiation-induced variability, our allele-specific method detected thousands of quantitative trait loci (QTLs) that influenced gene expression, chromatin accessibility, and RNA splicing. On the basis of these detected QTLs, we estimate that recall-by-genotype studies that use iPSC-derived cells will require cells from at least 20-80 individuals to detect the effects of regulatory variants with moderately large effect sizes.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Receptoras Sensoriales/metabolismo , Diferenciación Celular/genética , Línea Celular , Cromatina/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Técnicas de Genotipaje , Humanos , Sitios de Carácter Cuantitativo , Empalme del ARN , Células Receptoras Sensoriales/citología , Análisis de Secuencia de ARN , Análisis de la Célula Individual
9.
J Physiol ; 595(8): 2661-2679, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28105664

RESUMEN

KEY POINTS: Voltage-gated sodium channels play a fundamental role in determining neuronal excitability. Specifically, voltage-gated sodium channel subtype NaV 1.7 is required for sensing acute and inflammatory somatic pain in mice and humans but its significance in pain originating from the viscera is unknown. Using comparative behavioural models evoking somatic and visceral pain pathways, we identify the requirement for NaV 1.7 in regulating somatic (noxious heat pain threshold) but not in visceral pain signalling. These results enable us to better understand the mechanisms underlying the transduction of noxious stimuli from the viscera, suggest that the investigation of pain pathways should be undertaken in a modality-specific manner and help to direct drug discovery efforts towards novel visceral analgesics. ABSTRACT: Voltage-gated sodium channel NaV 1.7 is required for acute and inflammatory pain in mice and humans but its significance for visceral pain is unknown. Here we examine the role of NaV 1.7 in visceral pain processing and the development of referred hyperalgesia using a conditional nociceptor-specific NaV 1.7 knockout mouse (NaV 1.7Nav1.8 ) and selective small-molecule NaV 1.7 antagonist PF-5198007. NaV 1.7Nav1.8 mice showed normal nociceptive behaviours in response to intracolonic application of either capsaicin or mustard oil, stimuli known to evoke sustained nociceptor activity and sensitization following tissue damage, respectively. Normal responses following induction of cystitis by cyclophosphamide were also observed in both NaV 1.7Nav1.8 and littermate controls. Loss, or blockade, of NaV 1.7 did not affect afferent responses to noxious mechanical and chemical stimuli in nerve-gut preparations in mouse, or following antagonism of NaV 1.7 in resected human appendix stimulated by noxious distending pressures. However, expression analysis of voltage-gated sodium channel α subunits revealed NaV 1.7 mRNA transcripts in nearly all retrogradely labelled colonic neurons, suggesting redundancy in function. By contrast, using comparative somatic behavioural models we identify that genetic deletion of NaV 1.7 (in NaV 1.8-expressing neurons) regulates noxious heat pain threshold and that this can be recapitulated by the selective NaV 1.7 antagonist PF-5198007. Our data demonstrate that NaV 1.7 (in NaV 1.8-expressing neurons) contributes to defined pain pathways in a modality-dependent manner, modulating somatic noxious heat pain, but is not required for visceral pain processing, and advocate that pharmacological block of NaV 1.7 alone in the viscera may be insufficient in targeting chronic visceral pain.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/deficiencia , Nociceptores/metabolismo , Dolor Visceral/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Capsaicina/toxicidad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Planta de la Mostaza/toxicidad , Canal de Sodio Activado por Voltaje NAV1.7/genética , Dolor Nociceptivo/inducido químicamente , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Nociceptores/efectos de los fármacos , Aceites de Plantas/toxicidad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Dolor Visceral/inducido químicamente , Dolor Visceral/genética
10.
PLoS One ; 11(4): e0152405, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27050761

RESUMEN

Human genetic studies show that the voltage gated sodium channel 1.7 (Nav1.7) is a key molecular determinant of pain sensation. However, defining the Nav1.7 contribution to nociceptive signalling has been hampered by a lack of selective inhibitors. Here we report two potent and selective arylsulfonamide Nav1.7 inhibitors; PF-05198007 and PF-05089771, which we have used to directly interrogate Nav1.7's role in nociceptor physiology. We report that Nav1.7 is the predominant functional TTX-sensitive Nav in mouse and human nociceptors and contributes to the initiation and the upstroke phase of the nociceptor action potential. Moreover, we confirm a role for Nav1.7 in influencing synaptic transmission in the dorsal horn of the spinal cord as well as peripheral neuropeptide release in the skin. These findings demonstrate multiple contributions of Nav1.7 to nociceptor signalling and shed new light on the relative functional contribution of this channel to peripheral and central noxious signal transmission.


Asunto(s)
Axones/fisiología , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Terminales Presinápticos/fisiología , Potenciales de Acción , Animales , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Células HEK293 , Humanos , Masculino , Ratones , Canal de Sodio Activado por Voltaje NAV1.7/fisiología , Técnicas de Placa-Clamp , Éteres Fenílicos/farmacología , Sulfonamidas/farmacología
11.
Mol Ther ; 22(8): 1530-1543, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24832007

RESUMEN

The generation of human sensory neurons by directed differentiation of pluripotent stem cells opens new opportunities for investigating the biology of pain. The inability to generate this cell type has meant that up until now their study has been reliant on the use of rodent models. Here, we use a combination of population and single-cell techniques to perform a detailed molecular, electrophysiological, and pharmacological phenotyping of sensory neurons derived from human embryonic stem cells. We describe the evolution of cell populations over 6 weeks of directed differentiation; a process that results in the generation of a largely homogeneous population of neurons that are both molecularly and functionally comparable to human sensory neurons derived from mature dorsal root ganglia. This work opens the prospect of using pluripotent stem-cell-derived sensory neurons to study human neuronal physiology and as in vitro models for drug discovery in pain and sensory disorders.


Asunto(s)
Ganglios Espinales/fisiología , Canales Iónicos/genética , Células Madre Pluripotentes/metabolismo , Células Receptoras Sensoriales/fisiología , Análisis de la Célula Individual , Compuestos de Anilina/farmacología , Diferenciación Celular , Células Cultivadas , Colforsina/farmacología , Furanos/farmacología , Regulación de la Expresión Génica , Humanos , Dolor/fisiopatología , Células Receptoras Sensoriales/citología
12.
J Cell Biol ; 184(4): 491-500, 2009 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-19237596

RESUMEN

The slow Wallerian degeneration (Wld(S)) protein protects injured axons from degeneration. This unusual chimeric protein fuses a 70-amino acid N-terminal sequence from the Ube4b multiubiquitination factor with the nicotinamide adenine dinucleotide-synthesizing enzyme nicotinamide mononucleotide adenylyl transferase 1. The requirement for these components and the mechanism of Wld(S)-mediated neuroprotection remain highly controversial. The Ube4b domain is necessary for the protective phenotype in mice, but precisely which sequence is essential and why are unclear. Binding to the AAA adenosine triphosphatase valosin-containing protein (VCP)/p97 is the only known biochemical property of the Ube4b domain. Using an in vivo approach, we show that removing the VCP-binding sequence abolishes axon protection. Replacing the Wld(S) VCP-binding domain with an alternative ataxin-3-derived VCP-binding sequence restores its protective function. Enzyme-dead Wld(S) is unable to delay Wallerian degeneration in mice. Thus, neither domain is effective without the function of the other. Wld(S) requires both of its components to protect axons from degeneration.


Asunto(s)
Axones/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nicotinamida-Nucleótido Adenililtransferasa/metabolismo , Animales , Ratones , Ratones Transgénicos
13.
FEBS J ; 276(5): 1208-20, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19175675

RESUMEN

Endoplasmic reticulum (ER)-associated degradation (ERAD) is a cell-autonomous process that eliminates large quantities of misfolded, newly synthesized protein, and is thus essential for the survival of any basic eukaryotic cell. Accordingly, the proteins involved and their interaction partners are well conserved from yeast to mammals, and Saccharomyces cerevisiae is widely used as a model system with which to investigate this fundamental cellular process. For example, valosin-containing protein (VCP) and its yeast homologue cell division cycle protein 48 (Cdc48p), which help to direct polyubiquitinated proteins for proteasome-mediated degradation, interact with an equivalent group of ubiquitin ligases in mouse and in S. cerevisiae. A conserved structural motif for cofactor binding would therefore be expected. We report a VCP-binding motif (VBM) shared by mammalian ubiquitin ligase E4b (Ube4b)-ubiquitin fusion degradation protein 2a (Ufd2a), hydroxymethylglutaryl reductase degradation protein 1 (Hrd1)-synoviolin and ataxin 3, and a related sequence in M(r) 78,000 glycoprotein-Amfr with slightly different binding properties, and show that Ube4b and Hrd1 compete for binding to the N-terminal domain of VCP. Each of these proteins is involved in ERAD, but none has an S. cerevisiae homologue containing the VBM. Some other invertebrate model organisms also lack the VBM in one or more of these proteins, in contrast to vertebrates, where the VBM is widely conserved. Thus, consistent with their importance in ERAD, evolution has developed at least two ways to bring these proteins together with VCP-Cdc48p. However, the differing molecular architecture of VCP-Cdc48p complexes indicates a key point of divergence in the molecular details of ERAD mechanisms.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Ciclo Celular/genética , Retículo Endoplásmico/metabolismo , Evolución Molecular , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas de Ciclo Celular/metabolismo , Secuencia Conservada , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Proteínas de Saccharomyces cerevisiae , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteína que Contiene Valosina
14.
Mol Cell Neurosci ; 38(3): 325-40, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18468455

RESUMEN

Wallerian degeneration slow (Wld(S)) mice express a chimeric protein that delays axonal degeneration. The N-terminal domain (N70), which is essential for axonal protection in vivo, binds valosin-containing protein (VCP) and targets both Wld(S) and VCP to discrete nuclear foci. We characterized the formation, composition and localization of these potentially important foci. Missense mutations show that the N-terminal sixteen residues (N16) of Wld(S) are essential for both VCP binding and targeting Wld(S) to nuclear foci. Removing N16 abolishes foci, and VCP binding sequences from ataxin-3 or HrdI restore them. In vitro, these puncta co-localize with proteasome subunits. In vivo, Wld(S) assumes a range of nuclear distribution patterns, including puncta, and its neuronal expression and intranuclear distribution is region-specific and varies between spontaneous and transgenic Wld(S) models. We conclude that VCP influences Wld(S) intracellular distribution, and thus potentially its function, by binding within the N70 domain required for axon protection.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Líquido Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Adenosina Trifosfatasas/análisis , Adenosina Trifosfatasas/genética , Animales , Animales Modificados Genéticamente , Química Encefálica/genética , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Células Cultivadas , Citoplasma/química , Citoplasma/metabolismo , Células HeLa , Humanos , Líquido Intracelular/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/genética , Células PC12 , Unión Proteica/fisiología , Ratas , Proteína que Contiene Valosina , Degeneración Walleriana/genética , Degeneración Walleriana/metabolismo
15.
Genome Biol ; 8(10): R217, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17927811

RESUMEN

BACKGROUND: Changes in gene expression are hallmarks of cellular differentiation. Sexual differentiation in fission yeast (Schizosaccharomyces pombe) provides a model system for gene expression programs accompanying and driving cellular specialization. The expression of hundreds of genes is modulated in successive waves during meiosis and sporulation in S. pombe, and several known transcription factors are critical for these processes. RESULTS: We used DNA microarrays to investigate meiotic gene regulation by examining transcriptomes after genetic perturbations (gene deletion and/or overexpression) of rep1, mei4, atf21 and atf31, which encode known transcription factors controlling sexual differentiation. This analysis reveals target genes at a genome-wide scale and uncovers combinatorial control by Atf21p and Atf31p. We also studied two transcription factors not previously implicated in sexual differentiation whose meiotic induction depended on Mei4p: Rsv2p induces stress-related genes during spore formation, while Rsv1p represses glucose-metabolism genes. Our data further reveal negative feedback interactions: both Rep1p and Mei4p not only activate specific gene expression waves (early and middle genes, respectively) but are also required for repression of genes induced in the previous waves (Ste11p-dependent and early genes, respectively). CONCLUSION: These data give insight into regulatory principles controlling the extensive gene expression program driving sexual differentiation and highlight sophisticated interactions and combinatorial control among transcription factors. Besides triggering simultaneous expression of gene waves, transcription factors also repress genes in the previous wave and induce other factors that in turn regulate a subsequent wave. These dependencies ensure an ordered and timely succession of transcriptional waves during cellular differentiation.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Fúngica de la Expresión Génica , Schizosaccharomyces/genética , Diferenciación Sexual/genética , Factores de Transcripción Activadores/metabolismo , Meiosis/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Schizosaccharomyces/fisiología , Proteínas de Schizosaccharomyces pombe/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...